US6136978A - Camptothecin analogs and methods of preparation thereof - Google Patents

Camptothecin analogs and methods of preparation thereof Download PDF

Info

Publication number
US6136978A
US6136978A US09/212,178 US21217898A US6136978A US 6136978 A US6136978 A US 6136978A US 21217898 A US21217898 A US 21217898A US 6136978 A US6136978 A US 6136978A
Authority
US
United States
Prior art keywords
group
camptothecin
mmol
hydroxy
nmr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US09/212,178
Other languages
English (en)
Inventor
Dennis P. Curran
Hubert Josien
David Bom
Thomas G. Burke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Pittsburgh
Original Assignee
University of Pittsburgh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/921,102 external-priority patent/US6150343A/en
Application filed by University of Pittsburgh filed Critical University of Pittsburgh
Priority to US09/212,178 priority Critical patent/US6136978A/en
Assigned to UNIVERSITY OF PITTSBURGH reassignment UNIVERSITY OF PITTSBURGH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BOM, DAVID, CURRAN, DENNIS P., JOSIEN, HUBERT
Priority to CA2353822A priority patent/CA2353822C/en
Priority to KR1020017007539A priority patent/KR100750693B1/ko
Priority to EP99965287A priority patent/EP1140948A4/en
Priority to CNB998157686A priority patent/CN1177850C/zh
Priority to AU31236/00A priority patent/AU777786B2/en
Priority to PCT/US1999/029937 priority patent/WO2000035924A1/en
Priority to NZ51221099A priority patent/NZ512210A/xx
Priority to JP2000588183A priority patent/JP4638987B2/ja
Publication of US6136978A publication Critical patent/US6136978A/en
Application granted granted Critical
Priority to US10/134,781 priority patent/US6743917B2/en
Priority to HK02105702.6A priority patent/HK1044156B/zh
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF PITTSBURGH
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF PITTSBURGH
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • C07F7/0812Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • C07F7/0812Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring
    • C07F7/0814Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring said ring is substituted at a C ring atom by Si
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/0825Preparations of compounds not comprising Si-Si or Si-cyano linkages
    • C07F7/083Syntheses without formation of a Si-C bond

Definitions

  • the present invention relates to novel compounds and methods of preparation thereof and, particularly, to silyl camptothecin derivatives or analogs and to methods of preparation of such silyl camptothecin analogs.
  • camptothecin has a fused ring system generally comprising a pyrrolo[3,4-b]quinoline system (rings ABC) fused to a 2-pyridone ring (ring D), which, in turn, is fused to a lactone ring (ring E).
  • rings ABC pyrrolo[3,4-b]quinoline system
  • ring D 2-pyridone ring
  • ring E lactone ring
  • Camptothecin belongs to the family of topoisomerase I poisons. See, for example, Froelich-Ammon, S. J. et al., J. Biol. Chem., 270, 21429 (1995). Research to date strongly suggests that this molecule acts by interfering with the unwinding of supercoiled DNA by the cellular enzyme topoisomerase I, an enzyme which is usually overexpressed in malignant cells. In the highly replicating cancer cells, this triggers a cascade of events leading to apoptosis and programmed death. See Slichenmyer, W. J. et al., J. Natl. Cancer Inst., 85, 271 (1993). Recent advances at the molecular pharmacology level are reviewed in Pommier, Y. et al., Proc. Natl. Acad. Sci. USA, 92, 8861 (1995).
  • Camptothecin's initial clinical trials were limited by its poor solubility in physiologically compatible media.
  • early attempts to form a water-soluble sodium salt of camptothecin by opening the lactone ring with sodium hydroxide resulted in a compound having a poor antitumor activity. It was later reported that the closed lactone-form is an absolute requisite for antitumor activity. See Wani, M. C. et al., J. Med. Chem., 23, 554 (1980). More recently, structure-activity studies have identified analogous compounds with better solubility and better antitumor activity.
  • TPT topotecan
  • IRT irinotecan
  • GI-147211C GI-147211C is in late stage clinical trials.
  • These analogs are effective against a variety of refractory solid tumors such as malignant melanoma, stomach, breast, ovarian, lung and colorectal cancers, and seem particularly promising for the treatment of slow-dividing cancer lines. See, for example, Kingsbury, W. D. et al., J. Med. Chem., 34, 98 (1991); Sawada, S. et al., Chem. Pharm. Bull., 39, 1446 (1991); Luzzio, M. J. et al., J. Med.
  • camptothecin analogs that combine good to excellent anti-tumor activities with different solubility and biodistribution profiles could play a crucial role in the therapeutic arsenal for the treatment of various types of cancers.
  • camptothecin Given the proven beneficial biological activity of camptothecin and analogs thereof, it is desirable to develop additional camptothecin analogs and methods of preparation of camptothecin analogs.
  • the present invention provides generally a compound having the following formula (1): ##STR3##
  • the present invention also provides a method of synthesizing compounds having the formula (2): ##STR4## via a 4+1 radical annulation/cyclization wherein the precursor ##STR5## is reacted with an aryl isonitrile having the formula ##STR6##
  • R 1 and R 2 are independently the same or different and are preferably hydrogen, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, an aryloxy group, an acyloxy group, --OC(O)OR d , wherein R d is an alkyl group, a carbamoyloxy group, a halogen, a hydroxy group, a nitro group, a cyano group, an azido group, a formyl group, a hydrazino group, an acyl group (--C(O)R f wherein R f is preferably an alkyl group, an alkoxy group, an amino group or a hydroxy group), an amino group, --SR c , wherein, R c is hydrogen, an acyl group, an alkyl group, or an aryl group, or R 1 and R 2 together form a group of the formula --O(CH 2 ) n O-- wherein n represents the integer 1
  • R 3 is preferably H, a halogen, a nitro group, an amino group, a hydroxy group, or a cyano group.
  • R 2 and R 3 can also together form a group of the formula --O(CH 2 ) n O-- wherein n represents the integer 1 or 2.
  • R 4 is preferably H, F, a trialkylsilyl group, a C 1-3 alkyl group, a C 2-3 alkenyl group, a C 2-3 alkynyl group, or a C 1-3 alkoxy group.
  • R 5 is preferably a C 1-10 alkyl group.
  • a preferred alkyl group is an ethyl group.
  • Preferred substituted alkyl groups for R 5 include an allyl group, a propargyl and a benzyl group.
  • R 6 , R 7 and R 8 preferably are independently (the same or different) a C 1-10 alkyl group, a C 2-10 alkenyl group, a C 2-10 alkynyl group, or an aryl group.
  • a preferred substituted alkyl group for R 6 , R 7 and R 8 is a --(CH 2 ) N R 9 group, wherein N is an integer within the range of 1 through 10 and R 9 is a hydroxy group, an alkoxy group, an amino group, a halogen atom, a cyano group or a nitro group.
  • Preferred amino groups for R 9 include alkylamino groups and a dialkylamino groups.
  • R 11 is preferably an alkylene group, an alkenylene or an alkynylene group.
  • R 12 is preferably --CH ⁇ CH--CH 2 -- or --C.tbd.C--CH 2 --.
  • X is preferably Cl, Br or I. More preferably, X is Br or I. Most preferably, X is Br.
  • the present invention also provides a compound having the formula (2): ##STR7## wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 and R 11 are as defined prior to this paragraph.
  • the present invention further provides a compound of the above formula wherein one of R 1 , R 2 , R 3 , and R 4 is not H.
  • the present invention still further provides a compound of the above formula wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 and R 11 are as defined prior to this paragraph and wherein R 5 is a methyl group, a C 3-10 alkyl group, an allyl group, a benzyl group or a propargyl group.
  • the present invention further provides a compound having the following formula (3): ##STR8##
  • the present invention further provides a compound having the following formula (4): ##STR9##
  • alkyl refer generally to both unsubstituted and substituted groups unless specified to the contrary. Unless otherwise specified, alkyl groups are hydrocarbon groups and are preferably C 1 -C 15 (that is, having 1 to 15 carbon atoms) alkyl groups, and more preferably C 1 -C 10 alkyl groups, and can be branched or unbranched, acyclic or cyclic. The above definition of an alkyl group and other definitions apply also when the group is a substituent on another group (for example, an alkyl group as a substituent of an alkylamino group or a dialkylamino group).
  • aryl refers to phenyl or napthyl.
  • alkoxy refers to --OR d , wherein R d is an alkyl group.
  • aryloxy refers to --OR e , wherein R e is an aryl group.
  • acyl refers to --C(O)R f .
  • alkenyl refers to a straight or branched chain hydrocarbon group with at least one double bond, preferably with 2-15 carbon atoms, and more preferably with 3-10 carbon atoms (for example, --CH ⁇ CHR g ).
  • alkynyl refers to a straight or branched chain hydrocarbon group with at least one triple bond, preferably with 2-15 carbon atoms, and more preferably with 3-10 carbon atoms (for example, --C.tbd.CR h ).
  • alkylene alkenylene
  • alkynylene refer to bivalent forms of alkyl, alkenyl and alkynyl groups, respectively.
  • alkyl groups can be substituted with a wide variety of substituents to synthesize camptothecin analogs retaining activity.
  • alkyl groups may preferably be substituted with a group or groups including, but not limited to, a benzyl group, a phenyl group, an alkoxy group, a hydroxy group, an amino group (including, for example, free amino groups, alkylamino, dialkylamino groups and arylamino groups), an alkenyl group, an alkynyl group and an acyloxy group.
  • R a and R b are preferably independently hydrogen, an acyl group, an alkyl group, or an aryl group.
  • Acyl groups may preferably be substituted with (that is, R f is) an alkyl group, a haloalkyl group (for example, a perfluoroalkyl group), an alkoxy group, an amino group and a hydroxy group.
  • Alkynyl groups and alkenyl groups may preferably be substituted with (that is, R g and R h are preferably) a group or groups including, but not limited to, an alkyl group, an alkoxyalkyl group, an amino alkyl group and a benzyl group.
  • acyloxy refers to the group --OC(O)R d .
  • alkoxycarbonyloxy refers to the group --OC(O)OR d .
  • Amino and hydroxy groups may include protective groups as known in the art.
  • Preferred protective groups for amino groups include tert-butyloxycarbonyl, formyl, acetyl, benzyl, p-methoxybenzyloxycarbonyl, trityl.
  • Other suitable protecting groups as known to those skilled in the art are disclosed in Greene, T., Wuts, P. G. M., Protective Groups in Organic Synthesis, Wiley (1991), the disclosure of which is incorporated herein by reference.
  • R 1 , R 2 , R 3 , R 6 , R 7 and R 8 are preferably not excessively bulky to maintain activity of the resultant camptothecin analog.
  • R 1 , R 2 , R 3 , R 6 , R 7 and R 8 independently have a molecular weight less than approximately 250. More preferably R 1 , R 2 , R 3 , R 6 , R 7 and R 8 independently have a molecular weight less than approximately 200.
  • camptothecin analogs of the present invention can be prepared for pharmaceutical use as salts with inorganic acids such as, but not limited to, hydrochloride, hydrobromide, sulfate, phosphate, and nitrate.
  • the camptothecin analogs can also be prepared as salts with organic acids such as, but not limited to, acetate, tartrate, fumarate, succinate, citrate, methanesulfonate, p-toluenesulfonate, and stearate.
  • Other acids can be used as intermediates in the preparation of the compounds of the present invention and their pharmaceutically acceptable salts.
  • the E-ring (the lactone ring) may be opened with alkali metal such as, but not limited to, sodium hydroxide or calcium hydroxide, to form opened E-ring analogs of compounds of formula (1) as set forth in the compounds of formula (4).
  • alkali metal such as, but not limited to, sodium hydroxide or calcium hydroxide
  • the intermediates thus obtained are more soluble in water and may be purified to produce, after treatment with an acid, a purified form of the camptothecin analogs of the present invention.
  • the E-ring may also be modified to produce analogs of compounds of formula (1) with different solubility profiles in water or other solvents.
  • Methods to achieve this goal include, but are not limited to, opening the E-ring with hydroxide or a water-soluble amino group or functionalizing the hydroxy group at position 20 of the E-ring with a water-soluble group such as a polyethylene glycol group.
  • the analogs thus prepared act as pro-drugs. In other words, these analogs regenerate the compounds of formula (1) (with the closed E-ring structure) when administered to a living organism. See, Greenwald, R. B. et al., J. Med. Chem., 39, 1938 (1996).
  • analogs of the present invention are highly lipophilic and have been shown to enhance activity both in vivo and in vitro. Moreover, their A-ring substitution(s) have been shown to enhance blood stability.
  • the present invention also provides a method of treating a patient, which comprises administering a pharmaceutically effective amount of a compound of formulas (1) and/or (2) or a pharmaceutically acceptable salt thereof.
  • the compound may, for example, be administered to a patient afflicted with cancer and/or leukemia by any conventional route of administration, including, but not limited to, intravenously, intramuscularly, orally, subcutaneously, intratumorally, intradermally, and parenterally.
  • the pharmaceutically effective amount or dosage is preferably between 0.01 to 60 mg of one of the compounds of formulas (1) and (2) per kg of body weight. More preferably, the pharmaceutically effective amount or dosage is preferably between 0.1 to 40 mg of one of the compounds of formulas (1) and (2) per kg of body weight.
  • a pharmaceutically effective amount or dosage contains an amount of one of the compounds of formulas (1) and (2) effective to display antileukemic and/or antitumor (anticancer) behavior.
  • Pharmaceutical compositions containing as an active ingredient of one of the compounds of formulas (1) and (2) or a pharmaceutically acceptable salt thereof in association with a pharmaceutically acceptable carrier or diluent are also within the scope of the present invention.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising any of the compounds of formulas (1) and (2) and a pharmaceutically acceptable carrier.
  • the composition may, for example, contain between 0.1 mg and 3 g, and preferably between approximately 0.1 mg and 500 mg of the compounds of formulas (1), (2) and/or (4), and may be constituted into any form suitable for the mode of administration.
  • FIG. 1 is an illustration of a general synthetic scheme for the preparation of compounds of formula (1).
  • FIG. 2 is an illustration of a synthesis of (20S)-11-fluoro-7-trimethylsilylcamptothecin.
  • FIG. 3 is an illustration of a synthesis of (20S) -10-acetoxy-7-trimethylsilylcamptothecin and (20S) -10-hydroxy-7-trimethylsilylcamptothecin.
  • FIG. 4 is an illustration of a synthesis of (20S)-10-amino-7-trimethylsilylcamptothecin.
  • FIG. 5 is an illustration of a synthesis of (20S)-10-amino-11-fluoro-7-trimethylsilylcamptothecin.
  • FIG. 6 is an illustration of a synthesis of a novel analog of irinotecan.
  • FIG. 7 is an illustration of three representative silylcamptothecin analogs of formula (2).
  • FIG. 8 is an illustration of the synthesis of a propargyl bromide precursor.
  • FIG. 9 is an illustration of the synthesis of a radical precursor of formula (3).
  • FIG. 10 is an illustration of the reaction of the radical precursor of FIG. 9 with three isonitriles.
  • FIG. 11 is an illustration of the final step of the synthesis of the representative silylcamptothecin analogs of FIG. 7.
  • FIG. 12 is an illustration of excitation and emission fluorescence spectra of 1 ⁇ M (20S)-7-[(2-trimethylsilyl)ethyl]camptothecin, 7-TMSEt CPT, (36c) (DB-172).
  • FIG. 13 is an illustration of emission fluorescence spectra of 1 ⁇ M (20S)-10-amino-7-[(2-trimethylsilyl)ethyl]camptothecin, 10-NH2-7-TMSEt CPT (DB-173).
  • FIG. 14 is an illustration of emission fluorescence spectra of 1 ⁇ M (20S)-10-hydroxy-7-[(2-trimethylsilyl)ethyl]camptothecin, 10-OH-7-TMSEt CPT (DB-174).
  • FIG. 15 is an illustration of fluorescence spectra of 1 ⁇ M (20S)-10-hydroxy-7(tert-butyldimethylsilyl)camptothecin, 10-OH-7-TBS CPT (DB-67) in ethanol, 0.29 M DMPG and PBS.
  • FIG. 16 is an illustration of equilibrium binding of camptothecin analogs to DMPC.
  • FIG. 17 is an illustration of equilibrium binding of highly lipophilic camptothecin analogs of the present invention to DMPC.
  • FIG. 18 is an illustration of equilibrium binding of highly lipophilic camptothecin analogs of the present invention to DMPG.
  • FIG. 19 is an illustration of double-reciprocal plots for the binding of highly lipophillic camptothecin analogs of the present invention to DMPC small unilamellar vesicles (SUVs) at 37° C.
  • SUVs small unilamellar vesicles
  • FIG. 20 is an illustration of double-reciprocal plots for the binding of highly lipophillic camptothecin analogs of the present invention to DMPG SUVs at 37° C.
  • FIG. 21 is an illustration of the stability of DB-172 in PBS buffer pH 7.4 at 37° C.
  • FIG. 22 is an illustration of the dependence of fluorescence intensity of DB-172 on time and drug concentration.
  • FIG. 23 is an illustration of the fluorescence intensity of DB-172 in dependence on time and concentration.
  • FIG. 24 is an illustration of the dependence of the total fluorescence intensity on time and pH for the carboxylate form of DB-172.
  • FIG. 25 is an illustration of the dependence of total fluorescence intensity on time for several camptothecin analogs of the present invention.
  • FIG. 26 is an illustration of the drug stability of several camptothecin analogs of the present invention in phosphate buffered saline (PBS) and human blood.
  • PBS phosphate buffered saline
  • FIG. 27 is an illustration of the drug stability of several camptothecin analogs of the present invention in PBS, whole blood and PBS/human serum albumin (HSA).
  • FIG. 28 is an illustration of the plasma concentration of DB-67 after oral dosage.
  • R 1 and R 2 are preferably and independently (the same or different) H, a hydroxy group, a halo group, an amino group, a nitro group, a cyano group, a C 1-3 alkyl group, a C 1-3 perhaloalkyl group, a C 1-3 alkenyl group, a C 1-3 alkynyl group, a C 1-3 alkoxy group, a C 1-3 aminoalkyl group, a C 1-3 alkylamino group, a C 1-3 dialkylamino group, or R 1 and R 2 together form a group of the formula --O(CH 2 ) n O-- wherein n represents the integer 1 or 2.
  • R 1 and R 2 are independently (the same or different) H, a methyl group, an amino group, a nitro group, a cyano group, a hydroxy group, a hydroxymethyl group, a methylamino group, a dimethylamino group, an ethylamino group, a diethylamino group, an aminomethyl group, a methylaminomethyl group, a dimethylaminomethyl group, and the like.
  • R 3 is preferably F, an amino group, or a hydroxy group.
  • R 4 is preferably H, a trialkylsilyl group or F.
  • R 5 is preferably an ethyl group.
  • R 6 , R 7 and R 8 are preferably independently (the same or different) a C 1-6 alkyl group, a phenyl group or a --(CH 2 ) N R 10 group, wherein N is an integer within the range of 1 through 6 and R 10 is a halogen or a cyano group.
  • the compounds of formula (1) of the present invention can be prepared according to the general synthetic scheme shown in FIG. 1.
  • an iodopyridone (2) is first N-alkylated with a propargyl derivative (3) to produce radical precursor (4). Radical precursor (4) then undergoes a radical cascade with arylisonitrile (5) to generate product (1).
  • the N-alkylation proceeds smoothly following optimized conditions. See Curran, D. P. et al., Tetrahedron Lett., 36, 8917 (1995), the disclosure of which is incorporated herein by reference.
  • the synthesis of iodopyridone (2) and the conditions of the radical cascade have been previously reported.
  • the propargylating agent (3) is readily prepared by the standard silylation of the dianion of propargyl alcohol with a suitable silylating agent R 6 R 7 R 8 SiX followed by conversion of the propargyl alcohol to a leaving group such as a bromide, iodide or sulfonate. See Curran, D. P. et al., Angew. Chem. Int. Ed. Engl., 34, 2683 (1995), the disclosure of which is incorporated herein by reference, and U.S. patent application Ser. No. 08,436,799, filed May 8, 1995, the disclosures of which are incorporated herein by reference.
  • various reagents can be used in the radical cascade including, but not limited to, hexamethylditin, hexamethyldisilane, or tetrakis(trimethylsilyl)silane.
  • the source of energy for this reaction can be a sun lamp or an ultraviolet lamp.
  • the temperature is preferably set between approximately 25 and 150° C. More preferably, the temperature is set at approximately 70° C.
  • solvents include benzene, toluene, acetonitrile, THF and tert-butanol. Also, there is very broad latitude in the choice of substituents on the alkyne and the isonitrile because of the mildness of the reaction conditions.
  • FIG. 2 illustrates an embodiment of a general synthetic scheme for the synthesis of (20S)-11-fluoro-7-trimethylsilylcamptothecin (12).
  • a problem in this synthetic scheme is to control the regioselectivity of the radical cascade when both ortho positions in the arylisonitrile are available for cyclization (that is, R 4 is H in the final compound of formula (1)).
  • One solution to this problem relies upon the introduction of a trimethylsilyl group on the aryl isonitrile, (e.g. 3-fluoro-2-trimethylsilylphenyl isonitrile (9)).
  • the trimethylsilyl substituent blocks one of the ortho sites of the isonitrile toward cyclization and can be removed after the cascade reaction by hydrodesilylation.
  • the selectivity proceeds further in the sense that only one of the trimethylsilyl groups is removed in the last step.
  • FIGS. 3 to 6 Other embodiments of the general synthetic scheme for the preparation of several novel camptothecin derivatives of formula (1) are illustrated in FIGS. 3 to 6, and in the Examples.
  • FIGS. 7 through 11 The preparation of the compounds of formula (2) is illustrated in FIGS. 7 through 11.
  • three representative, novel A,B ring substituted silylcamptothecin compounds (36a), (36b), and (36c) are illustrated in FIG. 7.
  • radical precursor (43) was prepared as illustrated in FIG. 9.
  • (42) was alkylated with (41) to give the desired radical precursor (43) in 74% yield.
  • Reaction of (43) with the respective isonitrile (44a) or (44b) gave the protected silylcamptothecin derivatives (45a) and (45b) in 55% and 56% yields, respectively, as illustrated in FIG. 10.
  • deprotection of (45a) with 2 equivalents of K 2 CO 3 in MeOH/H 2 O solution gave a 47% yield of the 10-hydroxy derivative (36a) as illustrated in FIG. 10.
  • treatment with trifluoroacetic acid in CH 2 Cl 2 converted (45b) to the 10-amino derivative (36b) in 65% yield.
  • the method of the present invention also provides ready synthesis of (20S)-7-[(2-trimethylsilyl)ethyl]camptothecin (36c, FIG. 7). Reaction of phenyl isonitrile with iodo pyridone (43) gives this derivative in 52% yield (FIG. 10).
  • the (20S)-7-[(2-trimethylsilyl)ethyl]camptothecin (36c) and (20S)-7-(2-trimethylsilyl)camptothecin (disclosed in U.S. patent application Ser. No. 08/436,799) structures have recently been described by Hausheer et al. in International Patent Application Publication Number WO 98/07727.
  • the present invention thus provides a short and efficient synthetic scheme well suited to known structure-activity relationships in the camptothecin family. Indeed, the biological activity of the camptothecin skeleton is generally intolerant or has very little tolerance to substituents other than at the 7 and/or 9-11 positions. Following synthesis, these substituents are introduced via the alkynylderivative (3) and arylisonitrile (5), respectively.
  • the compounds of the present invention exhibit good to excellent antitumor activity as compared to camptothecin (CPT) and irinotecan (IRT).
  • camptothecin derivatives were evaluated for their cytotoxic effects on the growth of HL-60 (human promyelocytic leukemic), 833K (human teratocarcinoma) and DC-3F (hamster lung) cells in vitro.
  • the cells were cultured in an initial density of 5 ⁇ 10 -4 cell/ml. They were maintained in a 5% CO 2 humidified atmosphere at 37° C. in RPMI-1640 media (GIBCO-BRL Grand Island, N.Y.) containing penicillin 100u/ml)/streptomycin (100 ⁇ g/ml) (GIBCO-BRL) and 10% heat inactivated fetal bovine serum.
  • the assay was performed in duplicate in 96-well microplates.
  • cytotoxicity of the compounds toward HL-60 cells following 72 hr incubation was determined by XTT-microculture tetrazolium assay. Scudiero, D. A., et al., Cancer Res., 48, 4827 (1988), the disclosure of which is incorporated herein by reference. 2',3'-bis(-methoxy-4-nitro-5-sulfheny)-5-[(phenylamino)carbonyl]-2H-tetrazolium hydroxide (XTT) was prepared at 1 mg/ml in prewarmed (37° C.) medium without serum.
  • Phenazine methosulfate (PMS) and fresh XTT were mixed together to obtain 0.075 mM PMS-XTT solution (25 ⁇ l of the stock 5 mM PMS was added per 5 ml of 1 mg/ml XTT). Fifty ⁇ l of this mixture was added to each well of the cell culture at the end of 72 hr incubation. After incubation at 37° C. for 4 hr., absorbance at 450 nm and 630 nm was measured with a microplate reader (EL340, Bio-Tek Instruments, Inc., Winooski, Vt.).
  • the cytotoxicity of the camptothecin compounds toward 833K teratocarcinoma solid tumor cells and DC-3F hamster lung cells was determined in 96-well microplates by a method described by Skehan et al. for measuring cellular protein content. Skehan et al., "New Colorometric Cytotoxicity Assay for Anticancer Drug Screening," J. Nat'l Cancer Inst., 82, 1107 (1990), the disclosure of which is incorporated herein by reference. Cultures were fixed with trichloroacetic acid and then stained for 30 minutes with 0.4% sulforhodamine B dissolved in 1% acetic acid.
  • the reaction mixture comprised Tris-HCl buffer 10 mM, pH7.5; PBR 322 supercoiled double stranded circular DNA (4363 base pairs, from Bochringer Mannheim Biochemicals) 0.125 ⁇ g/ml, drug (camptothecin or its derivatives) concentration at 1, 10 and 100 ⁇ M, in the presence of purified DNA topoisomerase I with final volume of 20 ⁇ l as described previously.
  • tumor cells (1 ⁇ 10 6 ) were inoculated subcutaneously on day 0 and treatment started on day 1, intraperitoneously twice daily for five days.
  • the grading of effects was as described above.
  • camptothecin derivatives of formula (1) tested for the antitumor cytotoxicity in vitro exhibited higher potency than camptothecin in one to three cell lines. Most of those compounds exhibiting higher antitumor cytotoxicity also exhibited higher potency in enhancing the DNA-topoisomerase I-mediated cleavage of PBR 322 DNA, or in inhibiting the DNA-topoisomerase I-mediated relaxation of PBR 322 DNA. These results suggest excellent correlation between the antitumor cytoxicity of the camptothecin compounds with their ability to inhibit the functions of DNA-topoisomerase I.
  • 7-trimethylsilyl camptothecin showed better activity than camptothecin against sarcoma 180 in B 6 D 2 F 1 mice at several equivalent doses in a dose dependent manner in terms of tumor volume reduction.
  • 7-trimethylsilyl-11-flouro camptothecin exhibited a similar antitumor effect to camptothecin in terms of tumor volume reduction at 4-fold lower doses than camptothecin.
  • 7-trimethylsilyl-11-flouro camptothecin is more efficacious than camptothecin in its antitumor effects in vivo.
  • camptothecin In phosphate buffered saline (PBS) at pH 7.4, frequency-domain fluorescence lifetime spectroscopy reveals that human serum albumin (HSA) preferentially binds the carboxylate form of camptothecin with a 200-fold higher affinity than the lactone form. These interactions result in camptothecin opening more rapidly and completely in the presence of HSA than in the absence of the protein.
  • HSA human serum albumin
  • camptothecin lactone opens rapidly and completely to the carboxylate form with a t 1/2 value of 11 min and an almost negligible % lactone at equilibrium value of 0.2%.
  • camptothecin displayed enhanced stability (t 1/2 value of 22 min and a % lactone at equilibrium value of 5.3%).
  • the enhanced stability of camptothecin lactone in human blood was found to be due to drug associations with the lipid bilayers of red blood cells. Camptothecin binds erythrocyte membranes, the drug localizes within the acyl chain region, and accordingly remains protected from hydrolysis.
  • HSA plays an important role in determining the relative human blood stabilities of the camptothecins.
  • camptothecin and 9-aminocamptothecin the protein acts as a sink for the carboxylate drug form, binding the opened ring species and thereby shifting the lactone-carboxylate equilibria to the carboxylate.
  • topotecan, CPT-11, and SN-38 no such preferential binding of the carboxylate drug form by HSA is observed.
  • HSA preferentially binds the lactone form of SN-38 which thereby promotes higher circulatory levels of this biologically active species.
  • camptothecin analogs of the present invention exhibit unique properties that result in the agents displaying improved human blood stabilities while maintaining high anticancer activities.
  • camptothecin analogs were in the 20(S) configuration and were of high purity (>98%) as determined by HPLC assays with fluorescence detection. All other agents were reagent grade and were used without further purification. High purity water provided by a Milli-Q UV PLUS purification system (Bedford, Mass.) was utilized in all experiments.
  • Drug Stock Solution Preparation.
  • Stock solutions of the drugs were prepared in dimethylsulfoxide (A.C.S. spectrophotometric grade, Aldrich, Milwaukee, Wis.) at a concentration of 2 ⁇ 10 -3 M and stored in dark at 4° C.
  • L- ⁇ -Dimyristoylphosphatidylcholine (DMPC) and L- ⁇ -dimyristoylphosphatidylglycerol (DMPG) were obtained from Avanti Polar Lipids, Alabaster, Ala., and were used without further purification. All other chemicals were reagent grade and were used without further purification.
  • Vesicle Preparation Small unilamellar vesicle (SUV) suspensions were prepared the day of an experiment by the method of Burke and Tritton, "The Structure Basis of Anthracycline Selectivity for Unilamellar Phophatidylcholine Vesicles: An Equilibrium Binoinl Study," Biochem 24:1768-1776 (1985). Briefly, stock lipid suspensions containing 200 mg/mL lipid in phosphate buffered saline (PBS, pH 7.4) were prepared by Vortex mixing for 5-10 min above the TM of the lipid.
  • PBS phosphate buffered saline
  • the lipid dispersions were then sonicated using a bath-type sonicator (Laboratory Supplies Co., Hicksville, N.Y.) for 3-4 h until they became optically clear. A decrease in pH from 7.4 to 6.8 was observed for the SUV preparations of DMPG; therefore, the pH of these SUV suspensions was adjusted to 7.4 using small quantities of 2.5 M NaOH in PBS, followed by additional sonication. Each type of vesicle suspension was annealed for 30 min at 37° C. and then used in an experiment.
  • Steady-state fluorescence measurements were obtained on a SLM Model 9850 spectrofluorometer with a thermostated cuvette compartment. This instrument was interfaced with an IBM PS/2 model 55 SX computer. Excitation and emission spectra were recorded with an excitation resolution of 8 nm and an emission resolution of 4 nm. In all cases spectra were corrected for background fluorescence and scatter from unlabeled lipids or from solvents by subtraction of the spectrum of a blank. Steady-state fluorescence intensity measurements were made in the absence of polarizers. Steady-state anisotropy (a) measurements were determined with the instrument in the "T-format" for simultaneous measurement of two polarized intensities.
  • polarizer orientation was checked using a dilute solution of glycogen in water.
  • Anisotropy measurements for camptothecins were conducted using exciting light of 370 nm and long pass filters on each emission channel in order to isolate the drug's fluorescence signal from background scatter and/or residual fluorescence. All emission filters were obtained from Oriel Corp (Stamford, Conn.). The combination of exciting light and emission filters allowed adequate separation of fluorescence from background signal. The contribution of background fluorescence, together with scattered light, was typically less than 1% of the total intensity. Since the lactone rings of camptothecin and related congeners undergo hydrolysis in aqueous medium with half-lives of approximately 20 min., all measurements were completed within the shortest possible time (ca. 0.5 to 1 min) after mixing the drug stock solution with thermally pre-equilibrated solutions such that the experiments were free of hydrolysis product.
  • Double-reciprocal plots of the binding isotherms ⁇ 1/(bound fraction of drug) vs. 1/[lipid] ⁇ were linear and K values were determined from their slopes by the method of linear least squares analysis.
  • SRB sulphorrhodamine B
  • FIGS. 12 through 15 depict the fluorescence excitation and emission spectra of several of the new camptothecin analogs.
  • FIG. 12 summarizes the excitation and emission spectra of 1 ⁇ M DB-172 in phosphate buffered saline solution. The figure indicates that upon introduction of lipid bilayers into the sample there is an increase in the fluorescence emission of the compound, indicative of an interaction between the drug and the membrane. Upon changing the solvent to ethanol the fluorescence also changes.
  • FIGS. 13 through 15 summarize the emission spectra of DB-173, DB-174, and DB-67, respectively, in the presence and absence of membranes. In each case there is a marked increase in fluorescence intensity as the drug partitions into the lipid bilayer.
  • DB-172 (20S)-7-[(2-trimethylsilyl)ethyl]camptothecin (36c); DB-173, (20S)-10-amino-7-[(2-trimethylsilyl)ethyl]camptothecin (36b); DB-174, (20S)-10-hydroxy-7-[(2-trimethylsilyl)ethyl]camptothecin (36a); DB-67, (20S)-10-hydroxy-7(tert-butyldimethylsilyl)camptothecin; DB-148, (20S)-7-(3-chloropropyldimethylsilyl)camptothecin; DB-158, (20S)-10-hydroxy-7-(3-chloropropyldimethylsilyl) camptothecin; DB-202, (20S)-7(tert-butyldimethylsilyl) camptothecin; CHJ-792, 10-amino-7-trifluoride (36c)-(
  • a steady-state fluorescence anisotropy (a) measurement is related to the rotational rate of the fluorescent molecule through the Perrin Equation:
  • a o is the limiting fluorescence anisotropy in the absence of depolarizing rotations
  • is the excited-state lifetime
  • is the rotational correlation time of the fluorophore.
  • camptothecin in PBS, glycerol, and methanol were examined at 37° C. The lifetime values were determined to be 4.7 ns, 3.8 ns, and 3.5 ns, respectively. Similarly, camptothecin's lifetime value when associated with DMPC bilayers were measured at 37° C., and the average value for membrane-bound drug was found to be 3.7 ns.
  • camptothecin's excited-state lifetime is relatively insensitive to alterations in microenvironment (e.g. a change in solvent or fluorophore relocation from an aqueous milieu to a phospholipid membrane).
  • alterations in microenvironment e.g. a change in solvent or fluorophore relocation from an aqueous milieu to a phospholipid membrane.
  • the Perrin equation indicates a direct relationship between a and ⁇ values will exist (that is, as the ⁇ value of the fluorescent compound increases, then so too does its steady-state anisotropy value).
  • Steady-state fluorescence anisotropy values of the camptothecin analogues are highly sensitive to solvent viscosity and to associations with small unilamellar lipid vesicles.
  • topotecan has an a value of 0.008 in PBS, but its a value increases 9-fold and 40-fold in the viscous solvents octanol and glycerol, respectively.
  • a 21-fold enhancement in the a value of camptothecin is observed upon binding of drug to vesicles composed of either DMPC or DMPG.
  • the method of fluorescence anisotropy titration was employed to study the equilibrium binding of camptothecin analogs with lipid bilayers. As described previously, the experiment consisted of determining the a values for a set of samples where the drug concentration in each was held constant (typically 1 or 2 ⁇ M), while the lipid concentration among the members of a set was varied from 0 to 0.29 M.
  • [A B ] represents the concentration of bound drug
  • [A F ] represents the concentration of free drug
  • [L] represents the total lipid concentration in the vesicle suspension.
  • K may be determined from the inverse of the slope of a double reciprocal plot. In such a double reciprocal plot (representative data are shown in FIGS. 19 and 20), 1/fraction of the total drug bound is plotted vs. 1/lipid concentration, with a y-intercept value of 1 (for a system displaying binding site homogeneity).
  • DMPC fluid-phase and electroneutral L- ⁇ -dimyristoylphosphatidylcholine
  • DMPG fluid-phase and negatively-charged L- ⁇ -dimyristoylphosphatidylglycerol
  • the compounds of the present invention are much more lipophilic than either camptothecin or topotecan.
  • the affinities of DB67 for membranes composed of DMPC or DMPG are 27-fold and 28-fold greater that the corresponding values for camptothecin.
  • DB172 and DB174 are some 100-fold and 90-fold more apt to bind DMPC membranes when compared with camptothecin.
  • DB173 is also highly lipophilic, displaying a K value for DMPC some 58-fold greater than that observed for camptothecin.
  • the novel compounds of the present invention listed in Table 3 were found to display the highest membrane affinities by far when compared against other, previous camptothecin analogs containing the same ⁇ -hydroxy- ⁇ -lactone ring system.
  • FIG. 21 summarizes the stability of DB172 in phosphate buffered saline (PBS) buffer, pH 7.4, at physiological temperature. Shown in the figure are plots of lactone fraction as a function of time for DB172 at different concentrations. Drug was added to solution from a concentrated DMSO stock solution such that the volumes of DMSO were very small (less than 0.5%) relative to the volume of water. The drug stability was found to be markedly dependent on the drug concentration added. At the more dilute drug concentrations the drug hydrolyzes as previously observed for other camptothecins containing the ⁇ -hydroxy- ⁇ -lactone moiety. At high drug concentrations marked stabilization of the lactone ring of DB-172 was observed, a finding which is not typically observed for other camptothecins.
  • PBS phosphate buffered saline
  • FIG. 22 summarizes the dependence of the fluorescence intensity of DB172 as a function of time and pH.
  • DB172 is added to solution as the lactone form.
  • pH 10 where the conditions are such that the lactone more readily hydrolyzes and forms carboxylate, a significant change in fluorescence intensity is observed. It appears that a pH 10 nonfluorescent micellular aggregates composed of lactone disassemble and form open-ring carboxylate forms that tend to exist in solution as monomeric fluorescent species.
  • FIG. 23 explores the fluorescence intensity of DB172 as a function of concentration. Following the addition of low concentration of lactone drug to solution, the change in fluorescence signal is the greatest whereas at high drug concentration (10 ⁇ M) the fluorescence intensity changes are minimal. It is believed that a low concentration the micellular aggregates of DB172 displaying reduced fluorescence can disassemble and form fluorescent carboxylate species, but at higher drug concentration the equilibrium favors that the agent remains in the aggregated or reduced fluorescence state.
  • FIG. 24 shows that when the carboxylate form of DB172 is added to solution at pH 10, no change in fluorescence signal is observed at pH 10 while at lower pH values where lactone can form the fluorescence intensity decreases with time. Once again this decrease in fluorescence that occurs at reduced pH appear to be due to the formation of lactone aggregates of reduced fluorescence quantum yield.
  • FIGS. 21 through 24 are consistent with the unusual ability of DB172 lactone to self-associate and form micelles at micromolar drug concentrations.
  • the micellular DB172 aggregates display a reduced fluorescence. If conditions allow for hydrolysis to occur such that carboxylate forms, there is an increase in the fluorescence intensity of the sample.
  • FIG. 26 compares the stabilities of several new camptothecin analogs in their free form in PBS buffer (Panel A) versus in whole blood (Panel B).
  • FIG. 27 summarizes the stability data for DB172, DB173 and DB174 in PBS buffer only (Panel A), PBS buffer containing physiologically relevant 30 mg/ml levels of HSA (Panel B), and human blood (Panel C). All experiments were carried out at physiological temperature.
  • the highly biologically-active and lipophilic compound, 7-t-butyldimethylsilyl-10-hydroxycamptothecin (DB-67), was found to display superior stability in human blood, with a t 1/2 of 130 min and a % lactone at equilibrium value of 30 (compare with % lactone at equilibrium values in whole human blood for 9-aminocamptothecin ( ⁇ 0.3%), camptothecin (5.3%), topotecan (8.6%), CPT-11 (21.0%), and SN-38 (19.5%)).
  • the stability data are summarized in Table 4.
  • the new DB67 agent was found to be 25-times more lipophilic than camptothecin, and its 10-hydroxy functionality was found to markedly aid in promoting stability in the presence of HSA.
  • DB67 may be an ideal candidate for the treatment of brain cancer. With intrinsic activity several-fold greater than camptothecin, DB67 displays very high equilibrium lactone levels in human blood, is not tightly bound to human albumin like camptothecin and 9-aminocamptothecin, and is highly lipophilic which should enable the agent to more readily cross the blood brain barrier.
  • FIG. 28 contains data which demonstrate that DB-67 is absorbed from the gastrointestinal tract.
  • DB-67 is absorbed from the gastrointestinal tract.
  • the stock contained DB67 at a concentration of 1.6 mg/ml.
  • 1 ml samples of blood were drawn and collected by heart puncturing. The samples were centrifuged for 3 min and stored frozen until analysis was carried out. Note that DB67 levels rise above the 40 ng/ml level with much of the drug persisting as active lactone drug.
  • the novel highly lipophilic analogs described here may be administered orally and will appear in the bloodstream following administration.
  • HSA human serum albumin
  • the drug-HSA interactions can be manipulated by drug structural modification: A 10-OH substitution decreases the affinity of drug for HSA approximately 20-fold and an additional 7-ethyl substitution further alters the binding in favor of the lactone form.
  • a 10-OH substitution decreases the affinity of drug for HSA approximately 20-fold and an additional 7-ethyl substitution further alters the binding in favor of the lactone form.
  • SRB Sulphorhodamine B
  • cytotoxicities of various camptothecins against MDA-MB-435 tumorigenic metastatic human breast cancer cells in the absence and presence of 1 mg/ml HSA are summarized in Table 5.
  • the cytotoxicity values for cells exposed to drug for 72 hrs. are summarized in Table 5.
  • HSA is able to strongly attenuate the IC 50 values of camptothecin, but the extent to which HSA modulates the cytotoxicities of the new highly lipophilic analogs is significantly reduced.
  • 1 mg/ml HSA had no effect on the cytotoxic activity of DB173.
  • DB173 displays a low nM potency against the human breast cancer cells.
  • the ability of the agents to remain potent even in the presence of albumin is potentially significant because of the great abundance of this protein throughout the blood and tissue of the body.
  • a silyl group or a silylyalkyl group for example, a trimethylsilyl group or a trimethylsilylethyl group
  • introduction of a silyl group or a silyalkyl group at position 7 of the camptothecin structure typically results in a compound with better anti-tumor activity and human blood stability than camptothecin (see, for example, the compound of Example 1 as compared to (20S)-CPT) and other previous camptothecin analogs. Dual substitution at the 7 and 10 positions is even more favorable (see, for example, compounds DB-173 and DB-174).
  • the silyl group or the silylalkyl group is also beneficial in the irinotecan series (see, for example, the compound of Example 6 as compared to irinotecan).
  • the anti-tumor activity remains essentially unchanged when a hydroxy group is introduced at position 10 of the compound of Example 1 to produce the compound of Example 5.
  • the compound of Example 6 is a relative of SN-38, the active metabolite of irinotecan. High activities were also observed in the present studies when a trimethylsilyl group was introduced in conjunction with a fluoro atom at position 11 (see, for example, the compound of Example 7), or a primary amine group at positions 10 or 11 (see, respectively, Examples 8 and 9). Introduction of a fluoro atom in position 12 also results in an analog only approximately 2 times less potent than camptothecin (see, Example 11 as compared to (20S)-CPT). This result is surprising considering the poor activity of the 12-substituted camptothecins reported previously in the literature.
  • novel camptothecin analogs of the present invention have unique biophysical and physiological properties. These highly lipophilic camptothecin analogs with B-ring modifications and A- and B- ring modifications display markedly improved ⁇ -hydroxy- ⁇ -lactone ring stability in human blood. The camptothecin analogs of the present invention also display oral bioavailibility and potent anticancer activity even in the presence of human serum albumin.
  • a mammal (human or animal) may thus be treated by a method which comprises the administration to the mammal of a pharmaceutically effective amount of a compound of formula (1) or a pharmaceutically acceptable salt thereof.
  • the condition of the mammal can thereby be improved.
  • the compounds of the present invention can be administered in a variety of dosage forms including, for example: parenterally (for example, intravenously, intradermally, intramuscularly or subcutaneously); orally (for example, in the form of tablets, lozengers, capsules, suspensions or liquid solutions); rectally or vaginally, in the form of a suppository; or topically (for example, as a paste, cream, gel or lotion).
  • parenterally for example, intravenously, intradermally, intramuscularly or subcutaneously
  • orally for example, in the form of tablets, lozengers, capsules, suspensions or liquid solutions
  • rectally or vaginally in the form of a suppository
  • topically for example, as a paste, cream, gel or lotion.
  • Optimal dosages to be administered may be determined by those skilled in the art and will vary with the particular compound of formula (1) to be used, the strength of the preparation, the mode of administration, the time and frequency of administration, and the advancement of the patient's condition. Additional factors depending on the particular patient will result in the need to adjust dosages. Such factors include patient age, weight, gender and diet. Dosages may be administered at once or divided into a number of smaller doses administered at varying intervals of time.
  • Example 1-(2) Following the procedure described in Example 1-(2), the compound prepared in Example 1-(1) (44.5 mg, 0.10 mmol), the compound prepared in (4) (93.9 mg, 0.3 mmol), and hexamethylditin (50 mg, 0.15 mmol) in dry benzene (1.5 mL) were irradiated for 9 hours at 70° with a 275 W GE sunlamp.
  • the reaction was evaporated, dissolved in MeOH with a few drops of DMSO to aid solubility and injected into a Waters reverse phase HPLC.
  • the conditions used to effect separation were as follows. A Waters 600E system controller with a Waters 490E Programmable multiwavelength detector, a Sargent Welch plotter and Waters C-18 25 ⁇ 10 cartridge columns were employed.
  • the isonitrile was prepared in 2 steps via classical Boc-protection followed by dehydration. See Einhorn, J. et al., Synlett, 37 (1991). A mixture of 4-aminoformanilide (1.71 g, 12.6 mmol), di-tert-butyl dicarbonate (2.87 g, 13.2 mmol) and NaHCO 3 (1.11 g, 13.2 mmol) in absolute EtOH (50 mL) was sonicated in a cleaning bath for 4 h. The final solution was filtered through a pad of Celite and concentrated to dryness. The residue was taken up in half brine (50 mL), extracted with AcOEt (6 ⁇ 30 mL) and dried (Na 2 SO 4 ).
  • the isonitrile was prepared in 2 steps following the same procedures as described in Example 9-(1).
  • the Boc-protection of 3-aminoformanilide (1.80 g, 13.2 mmol) provided, after flash-chromatography (CHCl 3 /MeOH 95:5), 2.65 g (85%) of 3-tert-butyloxycarbonylaminoformanilide, as a white solid.
  • Example 1-(2) Following the procedure described in Example 1-(2), the compound prepared in Example 1-(1) (44.5 mg, 0.10 mmol) and the compound prepared in Example (1) (65.5 mg, 0.3 mmol) afforded, after flash-chromatographies (CHCl 3 /MeOH 95:5; CHCl 3 /acetone 5:1), 23.1 mg (43%) of a slightly yellow oil.
  • Example 1-(2) Following the procedure described in Example 1-(2), the compound prepared in Example 1-(1) (66.8 mg, 0.15 mmol) and the compound described in Example (3) (110 mg, 0.50 mmol) provided, after flash-chromatographies (CHCl 3 /MeOH 96:4; CHCl 3 /acetone 10:1), 47.6 mg (57%) of a slightly yellow oil containing the above regioisomers:
  • Example (4) The compound prepared in Example (4) (41.3 mg, 0.0746 mmol) was deprotected following the conditions described in Example 9-(3). After workup, the crude was subjected to a flash-chromatography (CHCl 3 /acetone/MeOH 70:10:1.5) to provide, in order of elution, first 14.1 mg (42%) of the pure (20S)-10-amino-11-fluoro-7-trimethylsilyl-camptothecin, then a 15.2 mg of a c.a. 1:1 mixture of (20S)-10-amino-11-fluoro-7-trimethylsilylcamptothecin and (20S)-10-amino-9-fluoro-7-trimethylsilylcamptothecin.
  • iodopyridone 2 150 mg, 0.450 mmol was combined with dimethyl-(1S, 2S, 5S)7,7 dimethylnorpinylsilyl-2-propynyl bromide (281 mg, 0.90 mmol).
  • iodopyridone 2 (150 mg, 0.450 mmol) was combined with 3-cyanopropyldimethylsilyl-2-propynyl bromide (165 mg, 0.678 mmol), K 2 CO 3 (124 mg, 0.90 mmol), Bu 4 N + Br - (14.5 mg, 0.045 mmol), H 2 O (0.02 mL) and toluene (3.6 mL). This mixture was refluxed for 1 h.
  • This known compound was prepared by a modification of the procedure in Piers, E.; Gavai, A. V. J. Org. Chem. 1990, 55, 2374.
  • To a flame dried flask was added dry CH 2 Cl 2 (150 mL) and triphenylphosphine (16 g, 61.2 mmol). The temperature was lowered to 0° C. and CBr 4 (10.15 g, 30.6 mmol) was added portionwise. After 30 min, a solution of 3-trimethylsilylpropanal (2.0 g, 15.3 mmol) in dry CH 2 Cl 2 (20 ml) was added. After 1 h at 0° C., the reaction was diluted with ether and filtered through celite.
  • PPh 3 (1.76 g, 6.73 mmol) followed by dry CH 2 Cl 2 (60 mL). The mixture was placed in an ice bath and bromine (0.34 mL, 6.41 mmol) was added dropwise. A small amount of PPh 3 was added until the reaction went from yellow to clear in color. After 0.5 h at 0° C., 5-trimethylsilanylpent-2-yn-1-ol (1.0 g, 6.41 mmol) was dissolved in CH 2 Cl 2 (5 mL) and added dropwise.
  • reaction mixture was poured into a separatory funnel, diluted with pentane (250 mL) and extracted with H 2 O (100 mL) and sat. NaHCO 3 (100 mL). The organic layer was dried (MgSO 4 ), filtered and reduced in volume to 50 mL. The crude solution was chromatographed on a pad of silica gel with pentane (500 mL).
  • This compound showed activity inhibiting cell proliferation in several lines of glioma cells (U87, A172, SG388, T98G, LN-Z308) with median effective concentrations of 10-100 ng/ml.
  • Example 21-(4) a solution of the iodopyridone prepared in Example 21-(4) above (56.8 mg, 0.12 mmol) was reacted with 4-tert-butyloxycarbonylaminophenyl isonitrile (65.4 mg, 0.3 mmol).
  • camptothecin derivative prepared in (2) above (17.5 mg, 0.031 mmol) was dissolved in CH 2 Cl 2 (1 mL) and trifluoroacetic acid (0.25 mL) was added. After 3 h at 22° C., the mixture was poured into saturated NaHCO 3 (20 mL) and extracted with EtOAc (10 ⁇ 15 mL).

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
US09/212,178 1993-06-30 1998-12-15 Camptothecin analogs and methods of preparation thereof Expired - Fee Related US6136978A (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US09/212,178 US6136978A (en) 1993-06-30 1998-12-15 Camptothecin analogs and methods of preparation thereof
KR1020017007539A KR100750693B1 (ko) 1998-12-15 1999-12-15 캄프토테신 유사체 및 그의 제조 방법
NZ51221099A NZ512210A (en) 1998-12-15 1999-12-15 Camptothecin analogs useful for treating cancer and leukemia
JP2000588183A JP4638987B2 (ja) 1998-12-15 1999-12-15 カンプトテシン類似体及びその調製方法
EP99965287A EP1140948A4 (en) 1998-12-15 1999-12-15 CAMPTOTHECIN ANALOGUE AND METHOD FOR THE PRODUCTION THEREOF
CNB998157686A CN1177850C (zh) 1998-12-15 1999-12-15 喜树碱类似物及其制备方法
AU31236/00A AU777786B2 (en) 1998-12-15 1999-12-15 Camptothecin analogs and methods of preparation thereof
PCT/US1999/029937 WO2000035924A1 (en) 1998-12-15 1999-12-15 Camptothecin analogs and methods of preparation thereof
CA2353822A CA2353822C (en) 1998-12-15 1999-12-15 Camptothecin analogs and methods of preparation thereof
US10/134,781 US6743917B2 (en) 1993-06-30 2002-04-29 Camptothecin analogs and methods of preparation thereof
HK02105702.6A HK1044156B (zh) 1998-12-15 2002-08-05 喜樹碱類似物及其製備方法

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US8519093A 1993-06-30 1993-06-30
US43679995A 1995-05-08 1995-05-08
US08/921,102 US6150343A (en) 1993-06-30 1997-08-29 Camptothecin analogs and methods of preparation thereof
US09/212,178 US6136978A (en) 1993-06-30 1998-12-15 Camptothecin analogs and methods of preparation thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/921,102 Continuation-In-Part US6150343A (en) 1993-06-30 1997-08-29 Camptothecin analogs and methods of preparation thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US61396800A Continuation 1993-06-30 2000-07-11

Publications (1)

Publication Number Publication Date
US6136978A true US6136978A (en) 2000-10-24

Family

ID=22789889

Family Applications (1)

Application Number Title Priority Date Filing Date
US09/212,178 Expired - Fee Related US6136978A (en) 1993-06-30 1998-12-15 Camptothecin analogs and methods of preparation thereof

Country Status (10)

Country Link
US (1) US6136978A (zh)
EP (1) EP1140948A4 (zh)
JP (1) JP4638987B2 (zh)
KR (1) KR100750693B1 (zh)
CN (1) CN1177850C (zh)
AU (1) AU777786B2 (zh)
CA (1) CA2353822C (zh)
HK (1) HK1044156B (zh)
NZ (1) NZ512210A (zh)
WO (1) WO2000035924A1 (zh)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6410731B2 (en) 1999-04-09 2002-06-25 University Of Pittsburgh Camptothecin analogs and methods of preparation thereof
US6497896B2 (en) 2001-02-12 2002-12-24 Supergen, Inc. Method for administering camptothecins via injection of a pharmaceutical composition comprising microdroplets containing a camptothecin
US6509027B2 (en) 2001-02-12 2003-01-21 Supergen, Inc. Injectable pharmaceutical composition comprising coated particles of camptothecin
US6723853B2 (en) 2001-08-27 2004-04-20 University Of Pittsburgh Intermediates and methods of preparation of intermediates in the enantiomeric synthesis of (20R)homocamptothecins and the enantiomeric synthesis of (20R)homocamptothecins
US7064202B1 (en) 2003-05-12 2006-06-20 University Of Kentucky Research Foundation Camptothecin intermediates and prodrugs and methods of preparation thereof
US7064206B1 (en) 2003-05-12 2006-06-20 University Of Kentucky Research Foundation Highly lipophilic camptothecin intermediates and prodrugs and methods of preparation thereof
US20080269169A1 (en) * 1993-06-30 2008-10-30 Curran Dennis P Camptothecin analogs and methods of preparation thereof
US20080302941A1 (en) * 2007-06-06 2008-12-11 Matthews Stewart D Vehicle Parking Assistance Device and Method for Use of Same
US20090099166A1 (en) * 2007-10-16 2009-04-16 Bionumerik Pharmaceuticals, Inc. C10-substituted camptothecin analogs
WO2009051580A1 (en) 2007-10-16 2009-04-23 Bionumerik Pharmaceuticals, Inc. C7-substituted camptothecin analogs
US20120282261A1 (en) * 2011-05-06 2012-11-08 Bionumerik Pharmaceuticals, Inc. Deuterated analogs of (4S)-4-Ethyl-4-hydroxy-11-[2- (trimethylsilyl)ethyl]-1H-pyrano[3', 4':6,7] indolizino [1,2-b]quinoline-3,14(4H, 12H)-dione and methods of use thereof
WO2014078168A1 (en) * 2012-11-13 2014-05-22 Bionumerik Pharmaceuticals, Inc. Methods for the total chemical synthesis of enantiomerically-pure 7-(2'-trimethylsilyl) ethyl camptothecin

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUP0700124A2 (en) * 2001-10-25 2007-06-28 Takeda Chemical Industries Ltd Quinoline compound
CA2548543C (en) * 2003-12-17 2012-01-03 Bionumerik Pharmaceuticals, Inc. Process for making camptothecin derivatives
EP2197279B1 (en) * 2007-10-16 2017-01-18 Bionumerik Pharmaceuticals, Inc. C10-substituted camptothecin analogs
CN107964257B (zh) * 2016-10-20 2021-07-09 香港理工大学深圳研究院 喹啉鎓离子骨架结构有机染料化合物及其制备方法和应用
CN116813631A (zh) * 2021-12-16 2023-09-29 迈威(上海)生物科技股份有限公司 一种喜树碱类化合物及其偶联物

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5468859A (en) * 1993-06-10 1995-11-21 Smithkline Beecham Corporation Process for asymmetric total synthesis of camptothecin analogues
WO1998007727A1 (en) * 1996-08-19 1998-02-26 Bionumerik Pharmaceuticals, Inc. Highly lipophilic camptothecin derivatives
WO1998035940A1 (en) * 1997-02-14 1998-08-20 Bionumerik Pharmaceuticals, Inc. Highly lipophilic camptothecin derivatives

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5744605A (en) * 1993-06-30 1998-04-28 University Of Pittsburgh Intermediates in the synthesis of (+) camptothecin and related compounds and synthesis thereof
US6150343A (en) * 1993-06-30 2000-11-21 University Of Pittsburgh Camptothecin analogs and methods of preparation thereof
JPH09123996A (ja) * 1995-10-30 1997-05-13 Sanshin Ind Co Ltd 船外機のシフト駆動機構

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5468859A (en) * 1993-06-10 1995-11-21 Smithkline Beecham Corporation Process for asymmetric total synthesis of camptothecin analogues
US5700939A (en) * 1993-06-10 1997-12-23 Smithkline Beecham Corporation Intermediates useful in camptothecin synthesis
WO1998007727A1 (en) * 1996-08-19 1998-02-26 Bionumerik Pharmaceuticals, Inc. Highly lipophilic camptothecin derivatives
US5910491A (en) * 1996-08-19 1999-06-08 Bionumerik Pharmaceuticals, Inc. Highly lipophilic camptothecin derivatives
WO1998035940A1 (en) * 1997-02-14 1998-08-20 Bionumerik Pharmaceuticals, Inc. Highly lipophilic camptothecin derivatives

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
Curran, D.P. and Liu, H., "New 4+1 Radical Annulations--A Formal Total Synthesis of (+/-)-Camptothecin," J. Am. Chem Soc., 114, 5863-5864 (1992). Published Jul. 1, 1992.
Curran, D.P. and Liu, H., New 4 1 Radical Annulations A Formal Total Synthesis of ( / ) Camptothecin, J. Am. Chem Soc., 114, 5863 5864 (1992). Published Jul. 1, 1992. *
Curran, D.P. et al., "Cascade Radical Reactions of Isonitriles: A Second-Generation Synthesis of (20S)-Camptothecin, Topotecan, Irinotecan, and GI-147211C," Angew. Chem. Int. Ed, 34, 2683-2684 (1995). Published Jan. 5, 1996.
Curran, D.P. et al., "Recent Applications of Radical Reactions in Natural Product Synthesis," Pure Appl. Chem., 65, 1153-1159 (1993). Published Jun. 1993.
Curran, D.P. et al., Cascade Radical Reactions of Isonitriles: A Second Generation Synthesis of (20S) Camptothecin, Topotecan, Irinotecan, and GI 147211C, Angew. Chem. Int. Ed, 34, 2683 2684 (1995). Published Jan. 5, 1996. *
Curran, D.P. et al., Recent Applications of Radical Reactions in Natural Product Synthesis, Pure Appl. Chem., 65, 1153 1159 (1993). Published Jun. 1993. *
Curran, D.P., "The Camptothecins--A reborn Family of Antitumor Agents", J. Chin. Chem. Soc., 40, 1-6 (1993). Published Feb. 1993.
Curran, D.P., Liu, H.; Josien, H; Ko, S.B., "Tandem Radical Reactions of Isonitriles with 2-pyrdonyl and other aryl radicals: Scope and Limitations, and a First Generation Sunthesis of (+/-)-Camptothecin," Tetrahedron, 52, 11385-11404 (1996). Published Aug. 1996.
Curran, D.P., Liu, H.; Josien, H; Ko, S.B., Tandem Radical Reactions of Isonitriles with 2 pyrdonyl and other aryl radicals: Scope and Limitations, and a First Generation Sunthesis of ( / ) Camptothecin, Tetrahedron, 52, 11385 11404 (1996). Published Aug. 1996. *
Curran, D.P., The Camptothecins A reborn Family of Antitumor Agents , J. Chin. Chem. Soc., 40, 1 6 (1993). Published Feb. 1993. *
Josien, H. et al., "7-Silylcamptothecins (Silatecans): A New Family of Camptothecin Antitumor Agents," Bioorg. Med. Chem. Lett. 7, 3189-3295 (1997).
Josien, H. et al., "A General Synthetic Approach to the (20S)-Camptothecin Family of Antitumor Agents by a Regiocontrolled Cascade Radical Cyclization of Aryl Isonitriles," Chem. Eur. J. 4, 67-83 (1998). Published Jan. 1998.
Josien, H. et al., "Synthesis of (S)-mappicine and Mappicine Ketone Via Radical Cascade Reaction of Isonitirles," Tetrahedron, 53, 8881-8886 (1997). Published Jun. 30, 1997.
Josien, H. et al., 7 Silylcamptothecins (Silatecans): A New Family of Camptothecin Antitumor Agents, Bioorg. Med. Chem. Lett. 7, 3189 3295 (1997). *
Josien, H. et al., A General Synthetic Approach to the (20S) Camptothecin Family of Antitumor Agents by a Regiocontrolled Cascade Radical Cyclization of Aryl Isonitriles, Chem. Eur. J. 4, 67 83 (1998). Published Jan. 1998. *
Josien, H. et al., Synthesis of (S) mappicine and Mappicine Ketone Via Radical Cascade Reaction of Isonitirles, Tetrahedron, 53, 8881 8886 (1997). Published Jun. 30, 1997. *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080269169A1 (en) * 1993-06-30 2008-10-30 Curran Dennis P Camptothecin analogs and methods of preparation thereof
US7655640B2 (en) * 1993-06-30 2010-02-02 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Camptothecin analogs and methods of preparation thereof
US7538220B2 (en) 1999-04-09 2009-05-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Intermediates in the preparation of homocamptothecin analogs
US6809103B2 (en) 1999-04-09 2004-10-26 University Of Pittsburgh Camptothecin analogs and methods of preparation thereof
US7982041B2 (en) 1999-04-09 2011-07-19 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Camptothecin analogs and methods of preparation thereof
US6410731B2 (en) 1999-04-09 2002-06-25 University Of Pittsburgh Camptothecin analogs and methods of preparation thereof
US7220860B2 (en) 1999-04-09 2007-05-22 University Of Pittsburgh Camptothecin analogs and methods of preparation thereof
US20080027224A1 (en) * 1999-04-09 2008-01-31 Curran Dennis P Camptothecin analogs and methods of preparation thereof
US20090198061A1 (en) * 1999-04-09 2009-08-06 Curran Dennis P Camptothecin analogs and methods of preparation thereof
US6497896B2 (en) 2001-02-12 2002-12-24 Supergen, Inc. Method for administering camptothecins via injection of a pharmaceutical composition comprising microdroplets containing a camptothecin
US6509027B2 (en) 2001-02-12 2003-01-21 Supergen, Inc. Injectable pharmaceutical composition comprising coated particles of camptothecin
US6723853B2 (en) 2001-08-27 2004-04-20 University Of Pittsburgh Intermediates and methods of preparation of intermediates in the enantiomeric synthesis of (20R)homocamptothecins and the enantiomeric synthesis of (20R)homocamptothecins
US7064206B1 (en) 2003-05-12 2006-06-20 University Of Kentucky Research Foundation Highly lipophilic camptothecin intermediates and prodrugs and methods of preparation thereof
US7064202B1 (en) 2003-05-12 2006-06-20 University Of Kentucky Research Foundation Camptothecin intermediates and prodrugs and methods of preparation thereof
US20080302941A1 (en) * 2007-06-06 2008-12-11 Matthews Stewart D Vehicle Parking Assistance Device and Method for Use of Same
WO2009051580A1 (en) 2007-10-16 2009-04-23 Bionumerik Pharmaceuticals, Inc. C7-substituted camptothecin analogs
US20090099166A1 (en) * 2007-10-16 2009-04-16 Bionumerik Pharmaceuticals, Inc. C10-substituted camptothecin analogs
US7687497B2 (en) * 2007-10-16 2010-03-30 Bionumerik Pharmaceuticals, Inc. C10-substituted camptothecin analogs
EP2197879A1 (en) * 2007-10-16 2010-06-23 Bionumerik Pharmaceuticals, Inc. C7-substituted camptothecin analogs
EP2197879A4 (en) * 2007-10-16 2012-02-15 Bionumerik Pharmaceuticals Inc CAMPTOTHECIN ANALOGUES SUBSTITUTED IN C7
US20120282261A1 (en) * 2011-05-06 2012-11-08 Bionumerik Pharmaceuticals, Inc. Deuterated analogs of (4S)-4-Ethyl-4-hydroxy-11-[2- (trimethylsilyl)ethyl]-1H-pyrano[3', 4':6,7] indolizino [1,2-b]quinoline-3,14(4H, 12H)-dione and methods of use thereof
US8569265B2 (en) * 2011-05-06 2013-10-29 Bionumerik Pharmaceuticals, Inc. Deuterated analogs of (4S)-4-ethyl-4-hydroxy-11-[2- (trimethylsilyl)ethyl]-1H-pyrano[3′, 4′:6,7] indolizino [1,2-b]quinoline-3,14(4H, 12H)-dione and methods of use thereof
WO2014078168A1 (en) * 2012-11-13 2014-05-22 Bionumerik Pharmaceuticals, Inc. Methods for the total chemical synthesis of enantiomerically-pure 7-(2'-trimethylsilyl) ethyl camptothecin
EP2920191A4 (en) * 2012-11-13 2016-06-22 Bionumerik Pharmaceuticals Inc METHODS FOR THE TOTAL CHEMICAL SYNTHESIS OF ENANTIOMERICALLY PURE 7- (2'-TRIMETHYLSILYL) ETHYL CAMPTOTHECIN

Also Published As

Publication number Publication date
NZ512210A (en) 2004-12-24
CN1177850C (zh) 2004-12-01
WO2000035924A1 (en) 2000-06-22
JP2002532505A (ja) 2002-10-02
HK1044156A1 (en) 2002-10-11
KR20010101250A (ko) 2001-11-14
CA2353822A1 (en) 2000-06-22
EP1140948A4 (en) 2008-01-02
EP1140948A1 (en) 2001-10-10
CN1352646A (zh) 2002-06-05
JP4638987B2 (ja) 2011-02-23
AU3123600A (en) 2000-07-03
CA2353822C (en) 2011-05-31
KR100750693B1 (ko) 2007-08-22
AU777786B2 (en) 2004-10-28
HK1044156B (zh) 2005-04-08

Similar Documents

Publication Publication Date Title
US7538220B2 (en) Intermediates in the preparation of homocamptothecin analogs
US6136978A (en) Camptothecin analogs and methods of preparation thereof
US7655640B2 (en) Camptothecin analogs and methods of preparation thereof
US6743917B2 (en) Camptothecin analogs and methods of preparation thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF PITTSBURGH, PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CURRAN, DENNIS P.;JOSIEN, HUBERT;BOM, DAVID;REEL/FRAME:010005/0585

Effective date: 19990308

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF PITTSBURGH;REEL/FRAME:023594/0172

Effective date: 20091125

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF PITTSBURGH;REEL/FRAME:023607/0379

Effective date: 20091125

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20121024